Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Mol Psychiatry ; 28(2): 588-600, 2023 02.
Article in English | MEDLINE | ID: mdl-36357671

ABSTRACT

Posttraumatic stress disorder (PTSD) after the pandemic has emerged as a major neuropsychiatric component of post-acute COVID-19 syndrome, yet the current pharmacotherapy for PTSD is limited. The use of adrenergic drugs to treat PTSD has been suggested; however, it is hindered by conflicting clinical results and a lack of mechanistic understanding of drug actions. Our studies, using both genetically modified mice and human induced pluripotent stem cell-derived neurons, reveal a novel α2A adrenergic receptor (α2AAR)-spinophilin-cofilin axis in the hippocampus that is critical for regulation of contextual fear memory reconsolidation. In addition, we have found that two α2 ligands, clonidine and guanfacine, exhibit differential abilities in activating this signaling axis to disrupt fear memory reconsolidation. Stimulation of α2AAR with clonidine, but not guanfacine, promotes the interaction of the actin binding protein cofilin with the receptor and with the dendritic spine scaffolding protein spinophilin to induce cofilin activation at the synapse. Spinophilin-dependent regulation of cofilin is required for clonidine-induced disruption of contextual fear memory reconsolidation. Our results inform the interpretation of differential clinical observations of these two drugs on PTSD and suggest that clonidine could provide immediate treatment for PTSD symptoms related to the current pandemic. Furthermore, our study indicates that modulation of dendritic spine morphology may represent an effective strategy for the development of new pharmacotherapies for PTSD.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Animals , Humans , Mice , Actin Depolymerizing Factors/pharmacology , Adrenergic Agents/pharmacology , Clonidine/pharmacology , Fear/physiology , Induced Pluripotent Stem Cells/metabolism , Microfilament Proteins/metabolism , Receptors, Adrenergic, alpha-2/metabolism
2.
J Pharmacol Exp Ther ; 384(2): 306-314, 2023 02.
Article in English | MEDLINE | ID: mdl-36456195

ABSTRACT

Development of neurocognitive disorder in human immunodeficiency virus (HIV)-infected patients has been linked to dysregulation of dopamine by the HIV-1 transactivator of transcription (Tat) protein, a negative allosteric modulator of dopamine transporter (DAT). Using fast scan cyclic voltammetry, the present study determined the effects of in vivo Tat expression on dopamine release in the caudate putamen of inducible Tat transgenic (iTat-tg) mice and the impact of a novel DAT allosteric modulator, Southern Research Institute (SRI)-32743, on the Tat effect. We found that 7- or 14-day doxycycline (Dox)-induced Tat expression in iTat-tg mice resulted in a 2-fold increase in phasic but not tonic stimulated baseline dopamine release relative to saline control mice. To determine whether the Tat-induced increase in dopamine release is mediated by DAT regulation, we examined the effect of an in vitro applied DAT inhibitor, nomifensine, on the dopamine release. Nomifensine (1 nM-10 µM) concentration-dependently enhanced phasic stimulated dopamine release in both saline- and Dox-treated iTat-tg mice, while the magnitude of the nomifensine-mediated dopamine release was unchanged between saline and Dox treatment groups. A single systemic administration of SRI-32743 prior to animal sacrifice reversed the increased dopamine release in the baseline of phasic dopamine release and nomifensine-augmented dopamine levels in Dox-treated iTat-tg mice, while SRI-32743 alone did not alter baseline of dopamine release. These findings suggest that Tat expression induced an increase in extracellular dopamine levels by not only inhibiting DAT-mediated dopamine transport but also stimulating synaptic dopamine release. Thus, DAT allosteric modulators may serve as a potential therapeutic intervention for HIV infection-dysregulated dopamine system observed in HIV-1 positive individuals. SIGNIFICANCE STATEMENT: HIV infection-induced dysregulation of the dopaminergic system has been implicated in the development of neurocognitive impairments observed in HIV positive patients. Understanding the mechanisms underlying HIV-1 Tat protein-induced alteration of extracellular dopamine levels will provide insights into the development of molecules that can attenuate Tat interaction with targets in the dopaminergic system. Here, we determined whether Tat alters dopamine release and how the novel DAT allosteric modulator, SRI-32743, impacts dopamine neurotransmission to attenuate Tat-induced effects on extracellular dopamine dynamics.


Subject(s)
HIV Infections , HIV-1 , Humans , Mice , Animals , Mice, Transgenic , HIV-1/metabolism , Dopamine/metabolism , Trans-Activators/metabolism , Nomifensine/metabolism , Putamen/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , tat Gene Products, Human Immunodeficiency Virus
3.
Int J Mol Sci ; 24(5)2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36901952

ABSTRACT

Mucopolysaccharidosis I-Hurler (MPS I-H) is caused by the loss of α-L-iduronidase, a lysosomal enzyme that degrades glycosaminoglycans. Current therapies cannot treat many MPS I-H manifestations. In this study, triamterene, an FDA-approved, antihypertensive diuretic, was found to suppress translation termination at a nonsense mutation associated with MPS I-H. Triamterene rescued enough α-L-iduronidase function to normalize glycosaminoglycan storage in cell and animal models. This new function of triamterene operates through premature termination codon (PTC) dependent mechanisms that are unaffected by epithelial sodium channel activity, the target of triamterene's diuretic function. Triamterene represents a potential non-invasive treatment for MPS I-H patients carrying a PTC.


Subject(s)
Mucopolysaccharidosis I , Animals , Mucopolysaccharidosis I/genetics , Iduronidase , Triamterene , Codon, Nonsense , Diuretics , Glycosaminoglycans/metabolism
4.
Bioorg Med Chem Lett ; 64: 128696, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35318165

ABSTRACT

Arsenicals belong to the class of chemical warfare agents known as vesicants, which are highly reactive, toxic and cause robust inflammatory response. Cutaneous exposure to arsenicals causes a wide range of systemic organ damage, beginning with cutaneous injuries, and later manifest multi-organ damage and death. Thus, the development of suitable antidotes that can effectively block injury following exposure to these agents is of great importance. Bromodomain 4 (BRD4), a member of the bromodomain and extra terminal domain (BET) family, plays crucial role in regulating transcription of inflammatory, proliferation and cell cycle genes. In this context, the development of potent small molecule inhibitors of BRD4 could serve as potential antidotes for arsenicals. Herein, we describe the synthesis and biological evaluation of a series of compounds.


Subject(s)
Arsenicals , Anti-Inflammatory Agents/chemistry , Antidotes/pharmacology , Arsenicals/pharmacology , Arsenicals/therapeutic use , Nuclear Proteins/metabolism , Transcription Factors/metabolism
6.
Antimicrob Agents Chemother ; 65(9): e0024421, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34152810

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a reemerging alphavirus that can cause encephalitis resulting in severe human morbidity and mortality. Using a high-throughput cell-based screen, we identified a quinolinone compound that protected against VEEV-induced cytopathic effects. Analysis of viral replication in cells identified several quinolinone compounds with potent inhibitory activity against vaccine and virulent strains of VEEV. These quinolinones also displayed inhibitory activity against additional alphaviruses, such as Mayaro virus and Ross River virus, although the potency was greatly reduced. Time-of-addition studies indicated that these compounds inhibit the early-to-mid stage of viral replication. Deep sequencing and reverse genetics studies identified two unique resistance mutations in the nsP2 gene (Y102S/C; stalk domain) that conferred VEEV resistance on this chemical series. Moreover, introduction of a K102Y mutation into the nsP2 gene enhanced the sensitivity of chikungunya virus (CHIKV) to this chemical series. Computational modeling of CHIKV and VEEV nsP2 identified a highly probable docking alignment for the quinolinone compounds that require a tyrosine residue at position 102 within the helicase stalk domain. These studies identified a class of compounds with antiviral activity against VEEV and other alphaviruses and provide further evidence that therapeutics targeting nsP2 may be useful against alphavirus infection.


Subject(s)
Chikungunya virus , Encephalitis Virus, Venezuelan Equine , Quinolones , Animals , Antiviral Agents/pharmacology , Encephalitis Virus, Venezuelan Equine/genetics , Horses , Humans , Quinolones/pharmacology , Virus Replication
7.
Molecules ; 26(21)2021 11 05.
Article in English | MEDLINE | ID: mdl-34771099

ABSTRACT

The delta opioid receptor (DOR) is a crucial receptor system that regulates pain, mood, anxiety, and similar mental states. DOR agonists, such as SNC80, and DOR-neutral antagonists, such as naltrindole, were developed to investigate the DOR in vivo and as potential therapeutics for pain and depression. However, few inverse agonists and non-competitive/irreversible antagonists have been developed, and none are widely available. This leaves a gap in our pharmacological toolbox and limits our ability to investigate the biology of this receptor. Thus, we designed and synthesized the novel compounds SRI-9342 as an irreversible antagonist and SRI-45128 as an inverse agonist. These compounds were then evaluated in vitro for their binding affinity by radioligand binding, their functional activity by 35S-GTPγS coupling, and their cAMP accumulation in cells expressing the human DOR. Both compounds demonstrated high binding affinity and selectivity at the DOR, and both displayed their hypothesized molecular pharmacology of irreversible antagonism (SRI-9342) or inverse agonism (SRI-45128). Together, these results demonstrate that we have successfully designed new inverse agonists and irreversible antagonists of the DOR based on a novel chemical scaffold. These new compounds will provide new tools to investigate the biology of the DOR or even new potential therapeutics.


Subject(s)
Analgesics, Opioid/chemistry , Binding, Competitive , Drug Discovery , Receptors, Opioid, delta/chemistry , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/pharmacology , Chemistry Techniques, Synthetic , Drug Discovery/methods , Humans , Ligands , Molecular Structure , Protein Binding , Receptors, Opioid, delta/agonists , Structure-Activity Relationship
8.
Int J Mol Sci ; 19(5)2018 May 20.
Article in English | MEDLINE | ID: mdl-29783777

ABSTRACT

Wnt/ß-catenin signaling is upregulated in triple-negative breast cancer (TNBC) compared to other breast cancer subtypes and normal tissues. Current Wnt/ß-catenin inhibitors, such as niclosamide, target the pathway nonspecifically and exhibit poor pharmacokinetics/pharmacodynamics in vivo. Niclosamide targets other pathways, including mTOR, STAT3 and Notch. Novel benzimidazoles have been developed to inhibit Wnt/ß-catenin signaling with greater specificity. The compounds SRI33576 and SRI35889 were discovered to produce more cytotoxicity in TNBC cell lines than in noncancerous cells. The agents also downregulated Wnt/ß-catenin signaling mediators LRP6, cyclin D1, survivin and nuclear active ß-catenin. In addition, SRI33576 did not affect mTOR, STAT3 and Notch signaling in TNBC and noncancerous cells. SRI35889 inhibited mTOR signaling less in noncancerous than in cancerous cells, while not affecting STAT3 and Notch pathways. Compounds SRI32529, SRI35357 and SRI35361 were not selectively cytotoxic against TNBC cell lines compared to MCF10A cells. While SRI32529 inhibited Wnt/ß-catenin signaling, the compound also mitigated mTOR, STAT3 and Notch signaling. SRI33576 and SRI35889 were identified as cytotoxic and selective inhibitors of Wnt/ß-catenin signaling with therapeutic potential to treat TNBC in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Triple Negative Breast Neoplasms/metabolism , Wnt Signaling Pathway/drug effects , Antineoplastic Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Cell Line, Tumor , Humans , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
9.
Biochem J ; 473(8): 1027-35, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26846349

ABSTRACT

Historically, drugs used in the treatment of cancers also tend to cause damage to healthy cells while affecting cancer cells. Therefore, the identification of novel agents that act specifically against cancer cells remains a high priority in the search for new therapies. In contrast with normal cells, most cancer cells contain multiple centrosomes which are associated with genome instability and tumorigenesis. Cancer cells can avoid multipolar mitosis, which can cause cell death, by clustering the extra centrosomes into two spindle poles, thereby enabling bipolar division. Kinesin-like protein KIFC1 plays a critical role in centrosome clustering in cancer cells, but is not essential for normal cells. Therefore, targeting KIFC1 may provide novel insight into selective killing of cancer cells. In the present study, we identified a small-molecule KIFC1 inhibitor, SR31527, which inhibited microtubule (MT)-stimulated KIFC1 ATPase activity with an IC50 value of 6.6 µM. By using bio layer interferometry technology, we further demonstrated that SR31527 bound directly to KIFC1 with high affinity (Kd=25.4 nM). Our results from computational modelling and saturation-transfer difference (STD)-NMR experiments suggest that SR31527 bound to a novel allosteric site of KIFC1 that appears suitable for developing selective inhibitors of KIFC1. Importantly, SR31527 prevented bipolar clustering of extra centrosomes in triple negative breast cancer (TNBC) cells and significantly reduced TNBC cell colony formation and viability, but was less toxic to normal fibroblasts. Therefore, SR31527 provides a valuable tool for studying the biological function of KIFC1 and serves as a potential lead for the development of novel therapeutic agents for breast cancer treatment.


Subject(s)
Drug Discovery , Kinesins/antagonists & inhibitors , Kinesins/metabolism , Thiadiazoles/chemistry , Thiadiazoles/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Discovery/methods , Humans , Kinesins/chemistry , Protein Binding/physiology , Protein Structure, Secondary , Thiadiazoles/pharmacology
10.
Bioorg Med Chem Lett ; 26(9): 2129-32, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27013392

ABSTRACT

γ-Secretase is one of two proteases directly involved in the production of the amyloid ß-peptide (Aß), which is pathogenic in Alzheimer's disease. Inhibition of γ-secretase to suppress the production of Aß should not block processing of one of its alternative substrates, Notch1 receptors, as interference with Notch1 signaling leads to severe toxic effects. In the course of our studies to identify γ-secretase inhibitors with selectivity for APP over Notch, 1 [3-(benzyl(isopropyl)amino)-1-(naphthalen-2-yl)propan-1-one] was found to inhibit γ-secretase-mediated Aß production without interfering with γ-secretase-mediated Notch processing in purified enzyme assays. As 1 is chemically unstable, efforts to increase the stability of this compound led to the identification of 2 [naphthalene-2-carboxylic acid benzyl-isopropyl-amide] which showed similar biological activity to compound 1. Synthesis and evaluation of a series of amide analogs resulted in benzofuranyl amide analogs that showed promising Notch-sparing γ-secretase inhibitory effects. This class of compounds may serve as a novel lead series for further study in the development of γ-secretase inhibitors.


Subject(s)
Amides/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Benzofurans/pharmacology , Naphthalenes/pharmacology , Protease Inhibitors/pharmacology , Receptor, Notch1/metabolism , Amides/chemical synthesis , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Benzofurans/chemical synthesis , Benzylamines/chemical synthesis , Benzylamines/pharmacology , Humans , Microsomes, Liver/metabolism , Naphthalenes/chemical synthesis , Peptide Fragments/antagonists & inhibitors , Protease Inhibitors/chemical synthesis , Rats , Signal Transduction , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 26(9): 2133-7, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27020305

ABSTRACT

One therapeutic approach for Alzheimer's disease is to inhibit the cleavage of the amyloid precursor protein (APP) by γ-secretase. At the beginning of a series of studies from our laboratories, a series of novel γ-amino alcohols (1) were found to possess γ-secretase inhibitory activity and Notch-sparing effects. A new one-pot synthesis of γ-amino alcohols and the structure-activity relationship (SAR) of these analogs will be discussed.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Propanolamines/pharmacology , Protease Inhibitors/pharmacology , Receptor, Notch1/metabolism , Amino Alcohols/chemical synthesis , Amino Alcohols/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Humans , Microsomes, Liver/metabolism , Naphthalenes/chemical synthesis , Naphthalenes/pharmacology , Peptide Fragments/antagonists & inhibitors , Propanolamines/chemical synthesis , Protease Inhibitors/chemical synthesis , Rats , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 26(9): 2138-41, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27038496

ABSTRACT

In search for novel lead compounds as γ-secretase inhibitors, analogs of aminopyrido[2,3-d]pyrimidin-7-ones (I) were synthesized and evaluated for inhibitory effects on amyloid-ß-peptide production and cleavage of the Notch1 receptor mediated by γ-secretase. Selected pyridopyrimidines, such as 1, 8, 9, 10, 11 and 16 are γ-secretase inhibitors that did not have an effect on Notch1 receptor processing.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidines/pharmacology , Receptor, Notch1/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Humans , Microsomes, Liver/metabolism , Peptide Fragments/antagonists & inhibitors , Protease Inhibitors/chemical synthesis , Pyridones/chemical synthesis , Pyrimidines/chemical synthesis , Rats , Structure-Activity Relationship
13.
Neurotherapeutics ; 21(1): e00291, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38241154

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia and lacks highly effective treatments. Tau-based therapies hold promise. Tau reduction prevents amyloid-ß-induced dysfunction in preclinical models of AD and also prevents amyloid-ß-independent dysfunction in diverse disease models, especially those with network hyperexcitability, suggesting that strategies exploiting the mechanisms underlying Tau reduction may extend beyond AD. Tau binds several SH3 domain-containing proteins implicated in AD via its central proline-rich domain. We previously used a peptide inhibitor to demonstrate that blocking Tau interactions with SH3 domain-containing proteins ameliorates amyloid-ß-induced dysfunction. Here, we identify a top hit from high-throughput screening for small molecules that inhibit Tau-FynSH3 interactions and describe its optimization with medicinal chemistry. The resulting lead compound is a potent cell-permeable Tau-SH3 interaction inhibitor that binds Tau and prevents amyloid-ß-induced dysfunction, including network hyperexcitability. These data support the potential of using small molecule Tau-SH3 interaction inhibitors as a novel therapeutic approach to AD.


Subject(s)
Alzheimer Disease , tau Proteins , Humans , tau Proteins/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , High-Throughput Screening Assays
14.
Cancers (Basel) ; 15(13)2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37444413

ABSTRACT

Cancer cells are especially sensitive to perturbations in ribosome biogenesis as they rely on finely tuned protein homeostasis to facilitate their rapid growth and proliferation. While ribosome synthesis and cancer have a well-established relationship, ribosome biogenesis has only recently drawn interest as a cancer therapeutic target. In this study, we exploited the relationship between ribosome biogenesis and cancer cell proliferation by using a potent ribosome biogenesis inhibitor, RBI2 (Ribosome Biogenesis Inhibitor 2), to perturb cancer cell growth and viability. We demonstrate herein that RBI2 significantly decreases cell viability in malignant melanoma cells and breast cancer cell lines. Treatment with RBI2 dramatically and rapidly decreased ribosomal RNA (rRNA) synthesis, without affecting the occupancy of RNA polymerase I (Pol I) on the ribosomal DNA template. Next-generation RNA sequencing (RNA-seq) revealed that RBI2 and previously described ribosome biogenesis inhibitor CX-5461 induce distinct changes in the transcriptome. An investigation of the content of the pre-rRNAs through RT-qPCR revealed an increase in the polyadenylation of cellular rRNA after treatment with RBI2, constituting a known pathway by which rRNA degradation occurs. Northern blotting revealed that RBI2 does not appear to impair or alter rRNA processing. Collectively, these data suggest that RBI2 inhibits rRNA synthesis differently from other previously described ribosome biogenesis inhibitors, potentially acting through a novel pathway that upregulates the turnover of premature rRNAs.

15.
Neurosci Lett ; 806: 137237, 2023 05 29.
Article in English | MEDLINE | ID: mdl-37059218

ABSTRACT

There is as of yet no FDA-approved medication for methamphetamine use disorder. Although dopamine D3 receptor antagonists have been shown to be useful in reducing methamphetamine seeking in animal models their translation to the clinic has been hindered because currently tested compounds can produce dangerously high blood pressure. Thus, it is important to continue to explore other classes of D3 antagonists. We report here the effects of SR 21502, a selective D3 receptor antagonist, on cue-induced reinstatement (i.e., relapse) of methamphetamine-seeking in rats. In Experiment 1, rats were trained to self-administer methamphetamine under a fixed ratio schedule of reinforcement followed by extinction of the response. Then, animals were tested with one of several doses of SR 21502 on cue-induced reinstatement of responding. SR 21502 significantly reduced cue-induced reinstatement of methamphetamine-seeking. In Experiment 2, animals were trained to lever press for food under a PR schedule and tested with the lowest dose of SR 21502 that caused a significant reduction in Experiment 1. These animals responded on average 8 times more than the vehicle-treated rats in Experiment 1, eliminating the possibility that SR 21502-treated rats in Experiment 1 responded less because they were incapacitated. In summary, these data suggest that SR 21502 may selectively inhibit methamphetamine-seeking and may constitute a promising pharmacotherapeutic agent for methamphetamine or other drug use disorders.


Subject(s)
Methamphetamine , Rats , Animals , Methamphetamine/pharmacology , Cues , Extinction, Psychological , Reinforcement, Psychology , Dopamine Antagonists/pharmacology , Self Administration , Dose-Response Relationship, Drug
16.
Biochemistry ; 51(1): 307-15, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22129351

ABSTRACT

Polyphenolic compounds including a number of natural products such as resveratrol, curcumin, catechin derivatives, and nordihydroguaiaretic acid have effects on the assembly of Aß fibrils and oligomers as well as on fibril morphology. Based on a lead structure obtained from a screen of a small molecule diversity library, simple benzoic acid derivatives distinguished by the number and position of hydroxyls on the aromatic ring displayed different abilities to dissociate preformed biotinyl-Aß(1-42) oligomers. The 2,3-, 2,5-, and 3,4-dihydroxybenzoic acid (DHBA) isomers were active oligomer dissociators. The remaining DHBA isomers and the monohydroxy and unsubstituted benzoic acids were inactive and did not compete with the active compounds to block oligomer dissociation. None of the compounds blocked oligomer assembly, indicating that they do not interact with monomeric Aß to shift the oligomer-monomer equilibrium. Dissociating activity was not associated with quinone redox cycling capacity of the compounds. Gallic acid (3,4,5-trihydroxybenzoic acid) stabilized biotinyl-Aß(1-42) oligomers against intrinsic dissociation and blocked the effects of the active dissociators, independent of the concentration of dissociator. A model for the mechanism of action of the DHBA dissociators proposes that these compounds destabilize oligomer structure promoting progressive monomer dissociation rather than fissioning oligomers into smaller, but still macromolecular, species. Gallic acid blocks dissociation by stabilizing oligomers against this process.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Benzoic Acid/chemistry , Catechols/chemistry , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Biotinylation , Dopamine/analogs & derivatives , Dopamine/chemistry , Glutaral/chemistry , Humans , Hydroxybenzoates , Isomerism , Peptide Fragments/metabolism , Proteolysis , Solubility
17.
Antioxidants (Basel) ; 11(10)2022 Sep 23.
Article in English | MEDLINE | ID: mdl-36290611

ABSTRACT

Acute kidney injury (AKI) is a major public health concern with significant morbidity and mortality and no current treatments beyond supportive care and dialysis. Preclinical studies have suggested that heme-oxygenase-1 (HO-1), an enzyme that catalyzes the breakdown of heme, has promise as a potential therapeutic target for AKI. Clinical trials involving HO-1 products (biliverdin, carbon monoxide, and iron), however, have not progressed beyond the Phase ½ level. We identified small-molecule inducers of HO-1 that enable us to exploit the full therapeutic potential of HO-1, the combination of its products, and yet-undefined effects of the enzyme system. Through cell-based, high-throughput screens for induction of HO-1 driven by the human HO-1 promoter/enhancer, we identified two novel small molecules and broxaldine (an FDA-approved drug) for further consideration as candidate compounds exhibiting an Emax ≥70% of 5 µM hemin and EC50 <10 µM. RNA sequencing identified shared binding motifs to NRF2, a transcription factor known to regulate antioxidant genes, including HMOX1. In vitro, the cytoprotective function of the candidates was assessed against cisplatin-induced cytotoxicity and apoptosis. In vivo, delivery of a candidate compound induced HO-1 expression in the kidneys of mice. This study serves as the basis for further development of small-molecule HO-1 inducers as preventative or therapeutic interventions for a variety of pathologies, including AKI.

18.
Viruses ; 14(4)2022 04 15.
Article in English | MEDLINE | ID: mdl-35458546

ABSTRACT

HIV-1 Vpu targets the host cell proteins CD4 and BST-2/Tetherin for degradation, ultimately resulting in enhanced virus spread and host immune evasion. The discovery and characterization of small molecules that antagonize Vpu would further elucidate the contribution of Vpu to pathogenesis and lay the foundation for the study of a new class of novel HIV-1 therapeutics. To identify novel compounds that block Vpu activity, we have developed a cell-based 'gain of function' assay that produces a positive signal in response to Vpu inhibition. To develop this assay, we took advantage of the viral glycoprotein, GaLV Env. In the presence of Vpu, GaLV Env is not incorporated into viral particles, resulting in non-infectious virions. Vpu inhibition restores infectious particle production. Using this assay, a high throughput screen of >650,000 compounds was performed to identify inhibitors that block the biological activity of Vpu. From this screen, we identified several positive hits but focused on two compounds from one structural family, SRI-41897 and SRI-42371. We developed independent counter-screens for off target interactions of the compounds and found no off target interactions. Additionally, these compounds block Vpu-mediated modulation of CD4, BST-2/Tetherin and antibody dependent cell-mediated toxicity (ADCC). Unfortunately, both SRI-41897 and SRI-42371 were shown to be specific to the N-terminal region of NL4-3 Vpu and did not function against other, more clinically relevant, strains of Vpu; however, this assay may be slightly modified to include more significant Vpu strains in the future.


Subject(s)
Anti-HIV Agents , HIV-1 , Human Immunodeficiency Virus Proteins , Viral Regulatory and Accessory Proteins , Viroporin Proteins , Anti-HIV Agents/chemistry , Bone Marrow Stromal Antigen 2/metabolism , GPI-Linked Proteins/metabolism , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/metabolism , Leukemia Virus, Gibbon Ape/metabolism , Small Molecule Libraries , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/metabolism , Viroporin Proteins/antagonists & inhibitors
19.
ACS Infect Dis ; 8(1): 91-105, 2022 01 14.
Article in English | MEDLINE | ID: mdl-34985256

ABSTRACT

HIV-1 Nef is an attractive target for antiretroviral drug discovery because of its role in promoting HIV-1 infectivity, replication, and host immune system avoidance. Here, we applied a screening strategy in which recombinant HIV-1 Nef protein was coupled to activation of the Src-family tyrosine kinase Hck, which enhances the HIV-1 life cycle in macrophages. Nef stimulates recombinant Hck activity in vitro, providing a robust assay for chemical library screening. High-throughput screening of more than 730 000 compounds using the Nef·Hck assay identified six unique hit compounds that bound directly to recombinant Nef by surface plasmon resonance (SPR) in vitro and inhibited HIV-1 replication in primary macrophages in the 0.04 to 5 µM range without cytotoxicity. Eighty-four analogs were synthesized around an isothiazolone scaffold from this series, many of which bound to recombinant Nef and inhibited HIV-1 infectivity in the low to submicromolar range. Compounds in this series restored MHC-I to the surface of HIV-infected primary cells and disrupted a recombinant protein complex of Nef with the C-terminal tail of MHC-I and the µ1 subunit of the AP-1 endocytic trafficking protein. Nef inhibitors in this class have the potential to block HIV-1 replication in myeloid cells and trigger recognition of HIV-infected cells by the adaptive immune system in vivo.


Subject(s)
HIV-1 , Down-Regulation , HIV-1/metabolism , Macrophages/metabolism , Virus Replication , src-Family Kinases/metabolism
20.
Neuropharmacology ; 220: 109239, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36126727

ABSTRACT

Cocaine abuse increases the incidence of HIV-1-associated neurocognitive disorders. We have demonstrated that HIV-1 transactivator of transcription (Tat) allosterically modulates dopamine (DA) reuptake through the human DA transporter (hDAT), potentially contributing to Tat-induced cognitive impairment and potentiation of cocaine conditioned place preference (CPP). This study determined the effects of a novel allosteric modulator of DAT, SRI-32743, on the interactions of HIV-1 Tat, DA, cocaine, and [3H]WIN35,428 with hDAT in vitro. SRI-32743 (50 nM) attenuated Tat-induced inhibition of [3H]DA uptake and decreased the cocaine-mediated dissociation of [3H]WIN35,428 binding in CHO cells expressing hDAT, suggesting a SRI-32743-mediated allosteric modulation of the Tat-DAT interaction. In further in vivo studies utilizing doxycycline-inducible Tat transgenic (iTat-tg) mice, 14 days of Tat expression significantly reduced the recognition index by 31.7% in the final phase of novel object recognition (NOR) and potentiated cocaine-CPP 2.7-fold compared to responses of vehicle-treated control iTat-tg mice. The Tat-induced NOR deficits and potentiation of cocaine-CPP were not observed in saline-treated iTat-tg or doxycycline-treated G-tg (Tat-null) mice. Systemic administration (i.p.) of SRI-32743 prior to behavioral testing ameliorated Tat-induced impairment of NOR (at a dose of 10 mg/kg) and the Tat-induced potentiation of cocaine-CPP (at doses of 1 or 10 mg/kg). These findings demonstrate that Tat and cocaine interactions with DAT may be regulated by compounds interacting at the DAT allosteric modulatory sites, suggesting a potential therapeutic intervention for HIV-infected patients with concurrent cocaine abuse.


Subject(s)
Cocaine-Related Disorders , Cocaine , HIV-1 , Animals , Cocaine/metabolism , Cocaine/pharmacology , Cocaine-Related Disorders/drug therapy , Cricetinae , Cricetulus , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Doxycycline , Humans , Mice , Mice, Transgenic , Reward , Trans-Activators , Transcription Factor DP1/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL